1
|
Zhang C, Ma Y, Zhao Y, Guo N, Han C, Wu Q, Mu C, Zhang Y, Tan S, Zhang J, Liu X. Systematic review of melatonin in cerebral ischemia-reperfusion injury: critical role and therapeutic opportunities. Front Pharmacol 2024; 15:1356112. [PMID: 38375039 PMCID: PMC10875093 DOI: 10.3389/fphar.2024.1356112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Cerebral ischemia-reperfusion (I/R) injury is the predominant causes for the poor prognosis of ischemic stroke patients after reperfusion therapy. Currently, potent therapeutic interventions for cerebral I/R injury are still very limited. Melatonin, an endogenous hormone, was found to be valid in preventing I/R injury in a variety of organs. However, a systematic review covering all neuroprotective effects of melatonin in cerebral I/R injury has not been reported yet. Thus, we perform a comprehensive overview of the influence of melatonin on cerebral I/R injury by collecting all available literature exploring the latent effect of melatonin on cerebral I/R injury as well as ischemic stroke. In this systematic review, we outline the extensive scientific studies and summarize the beneficial functions of melatonin, including reducing infarct volume, decreasing brain edema, improving neurological functions and attenuating blood-brain barrier breakdown, as well as its key protective mechanisms on almost every aspect of cerebral I/R injury, including inhibiting oxidative stress, neuroinflammation, apoptosis, excessive autophagy, glutamate excitotoxicity and mitochondrial dysfunction. Subsequently, we also review the predictive and therapeutic implications of melatonin on ischemic stroke reported in clinical studies. We hope that our systematic review can provide the most comprehensive introduction of current advancements on melatonin in cerebral I/R injury and new insights into personalized diagnosis and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chenguang Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yumei Ma
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yating Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shutong Tan
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Wei Z, Xie Y, Wei M, Zhao H, Ren K, Feng Q, Xu Y. New insights in ferroptosis: Potential therapeutic targets for the treatment of ischemic stroke. Front Pharmacol 2022; 13:1020918. [PMID: 36425577 PMCID: PMC9679292 DOI: 10.3389/fphar.2022.1020918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/26/2022] [Indexed: 10/22/2023] Open
Abstract
Stroke is a common disease in clinical practice, which seriously endangers people's physical and mental health. The neurovascular unit (NVU) plays a key role in the occurrence and development of ischemic stroke. Different from other classical types of cell death such as apoptosis, necrosis, autophagy, and pyroptosis, ferroptosis is an iron-dependent lipid peroxidation-driven new form of cell death. Interestingly, the function of NVU and stroke development can be regulated by activating or inhibiting ferroptosis. This review systematically describes the NVU in ischemic stroke, provides a comprehensive overview of the regulatory mechanisms and key regulators of ferroptosis, and uncovers the role of ferroptosis in the NVU and the progression of ischemic stroke. We further discuss the latest progress in the intervention of ferroptosis as a therapeutic target for ischemic stroke and summarize the research progress and regulatory mechanism of ferroptosis inhibitors on stroke. In conclusion, ferroptosis, as a new form of cell death, plays a key role in ischemic stroke and is expected to become a new therapeutic target for this disease.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Ye D, Xu Y, Shi Y, Fan M, Lu P, Bai X, Feng Y, Hu C, Cui K, Tang X, Liao J, Huang W, Xu F, Liang X, Huang J. Anti-PANoptosis is involved in neuroprotective effects of melatonin in acute ocular hypertension model. J Pineal Res 2022; 73:e12828. [PMID: 36031799 DOI: 10.1111/jpi.12828] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
Acute ocular hypertension (AOH) is the most important characteristic of acute glaucoma, which can lead to retinal ganglion cell (RGC) death and permanent vision loss. So far, approved effective therapy is still lacking in acute glaucoma. PANoptosis (pyroptosis, apoptosis, and necroptosis), which consists of three key modes of programmed cell death-apoptosis, necroptosis, and pyroptosis-may contribute to AOH-induced RGC death. Previous studies have demonstrated that melatonin (N-acetyl-5-methoxytryptamine) exerts a neuroprotective effect in many retinal degenerative diseases. However, whether melatonin is anti-PANoptotic and neuroprotective in the progression of acute glaucoma remains unclear. Thus, this study aimed to explore the role of melatonin in AOH retinas and its underlying mechanisms. The results showed that melatonin treatment attenuated the loss of ganglion cell complex thickness, retinal nerve fiber layer thickness, and RGC after AOH injury, and improved the amplitudes of a-wave, b-wave, and oscillatory potentials in the electroretinogram. Additionally, the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells was decreased, and the upregulation of cleaved caspase-8, cleaved caspase-3, Bax, and Bad and downregulation of Bcl-2 and p-Bad were inhibited after melatonin administration. Meanwhile, both the expression and activation of MLKL, RIP1, and RIP3, along with the number of PI-positive cells, were reduced in melatonin-treated mice, and p-RIP3 was in both RGC and microglia/macrophage after AOH injury. Furthermore, melatonin reduced the expression of NLRP3, ASC, cleaved caspase-1, gasdermin D (GSDMD), and cleaved GSDMD, and decreased the number of Iba1/interleukin-1β-positive cells. In conclusion, melatonin ameliorated retinal structure, prevented retinal dysfunction after AOH, and exerted a neuroprotective effect via inhibition of PANoptosis in AOH retinas.
Collapse
Affiliation(s)
- Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Peng Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Bai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanlin Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chenyang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Liao
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Wei Huang
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Fan Xu
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
4
|
Fang Y, Zhao C, Xiang H, Jia G, Zhong R. Melatonin improves cryopreservation of ram sperm by inhibiting mitochondrial permeability transition pore opening. Reprod Domest Anim 2020; 55:1240-1249. [DOI: 10.1111/rda.13771] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/04/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Yi Fang
- Jilin Provincial Key Laboratory of Grassland Farming Northeast Institute of Geography and Agoecology Chinese Academy of Sciences Changchun, Jilin China
| | - Chengzhen Zhao
- Jilin Provincial Key Laboratory of Grassland Farming Northeast Institute of Geography and Agoecology Chinese Academy of Sciences Changchun, Jilin China
| | - Hai Xiang
- Jilin Provincial Key Laboratory of Grassland Farming Northeast Institute of Geography and Agoecology Chinese Academy of Sciences Changchun, Jilin China
| | - GongXue Jia
- Key Laboratory of Adaptation and Evolution of Plateau Biota Northwest Institute of Plateau Biology Chinese Academy of Sciences Xining China
| | - Rongzhen Zhong
- Jilin Provincial Key Laboratory of Grassland Farming Northeast Institute of Geography and Agoecology Chinese Academy of Sciences Changchun, Jilin China
| |
Collapse
|
5
|
Tan HY, Ng KY, Koh RY, Chye SM. Pharmacological Effects of Melatonin as Neuroprotectant in Rodent Model: A Review on the Current Biological Evidence. Cell Mol Neurobiol 2020; 40:25-51. [PMID: 31435851 DOI: 10.1007/s10571-019-00724-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
The progressive loss of structure and functions of neurons, including neuronal death, is one of the main factors leading to poor quality of life. Promotion of functional recovery of neuron after injury is a great challenge in neuroregenerative studies. Melatonin, a hormone is secreted by pineal gland and has antioxidative, anti-inflammatory, and anti-apoptotic properties. Besides that, melatonin has high cell permeability and is able to cross the blood-brain barrier. Apart from that, there are no reported side effects associated with long-term usage of melatonin at both physiological and pharmacological doses. Thus, in this review article, we summarize the pharmacological effects of melatonin as neuroprotectant in central nervous system injury, ischemic-reperfusion injury, optic nerve injury, peripheral nerve injury, neurotmesis, axonotmesis, scar formation, cell degeneration, and apoptosis in rodent models.
Collapse
Affiliation(s)
- Hui Ying Tan
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia.
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Abstract
Brain injury in the full-term and near-term neonates is a significant cause of mortality and long-term morbidity, resulting in injury patterns distinct from that seen in premature infants and older patients. Therapeutic hypothermia improves long-term outcomes for many of these infants, but there is a continued search for therapies to enhance the plasticity of the newborn brain, resulting in long-term repair. It is likely that a combination strategy utilizing both early and late interventions may have the most benefit, capitalizing on endogenous mechanisms triggered by hypoxia or ischemia. Optimizing care of these critically ill newborns in the acute setting is also vital for improving both short- and long-term outcomes.
Collapse
|
7
|
Jin Y, Choi J, Lee S, Kim JW, Hong Y. Pathogenetical and Neurophysiological Features of Patients with Autism Spectrum Disorder: Phenomena and Diagnoses. J Clin Med 2019; 8:E1588. [PMID: 31581672 PMCID: PMC6832208 DOI: 10.3390/jcm8101588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 12/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that is accompanied by social deficits, repetitive and restricted interests, and altered brain development. The majority of ASD patients suffer not only from ASD itself but also from its neuropsychiatric comorbidities. Alterations in brain structure, synaptic development, and misregulation of neuroinflammation are considered risk factors for ASD and neuropsychiatric comorbidities. Electroencephalography has been developed to quantitatively explore effects of these neuronal changes of the brain in ASD. The pineal neurohormone melatonin is able to contribute to neural development. Also, this hormone has an inflammation-regulatory role and acts as a circadian key regulator to normalize sleep. These functions of melatonin may play crucial roles in the alleviation of ASD and its neuropsychiatric comorbidities. In this context, this article focuses on the presumable role of melatonin and suggests that this hormone could be a therapeutic agent for ASD and its related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yunho Jin
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae 50834, Korea.
| | - Jeonghyun Choi
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae 50834, Korea.
| | - Seunghoon Lee
- Gimhae Industry Promotion & Biomedical Foundation, Gimhae 50969, Korea.
| | - Jong Won Kim
- Department of Healthcare Information Technology, College of Bio-Nano Information Technology, Inje University, Gimhae 50834, Korea.
| | - Yonggeun Hong
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae 50834, Korea.
- Department of Medicine, Division of Hematology/Oncology, Harvard Medical School-Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|
8
|
Tiong YL, Ng KY, Koh RY, Ponnudurai G, Chye SM. Melatonin Prevents Oxidative Stress-Induced Mitochondrial Dysfunction and Apoptosis in High Glucose-Treated Schwann Cells via Upregulation of Bcl2, NF-κB, mTOR, Wnt Signalling Pathways. Antioxidants (Basel) 2019; 8:antiox8070198. [PMID: 31247931 PMCID: PMC6680940 DOI: 10.3390/antiox8070198] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Neuropathy is a complication that affects more than 50% of long-standing diabetic patients. One of the causes of diabetes neuropathy (DN) is the apoptosis of Schwann cells due to prolonged exposure to high glucose and build-up of oxidative stress. Melatonin is a hormone that has a known antioxidant property. In this study, we investigated the protective effect of melatonin on high glucose-induced Schwann cells' apoptosis. Our results revealed that high glucose promoted apoptosis via mitochondrial-related oxidative stress and downregulated Bcl-2 family proteins in Schwann cells. In this signalling pathway, Bcl-2, Bcl-XL and Mcl-1 proteins were down-regulated while p-BAD and Puma proteins were up-regulated by high glucose treatment. Besides, we also proved that high glucose promoted apoptosis in Schwann cells through decreasing the p-NF-κB in the NF-κB signalling pathway. Key regulators of mTOR signalling pathway such as p-mTOR, Rictor and Raptor were also down-regulated after high glucose treatment. Additionally, high glucose treatment also decreased the Wnt signalling pathway downstream proteins (Wnt 5a/b, p-Lrp6 and Axin). Our results showed that melatonin treatment significantly inhibited high glucose-induced ROS generation, restored mitochondrial membrane potential and inhibited high glucose-induced apoptosis in Schwann cells. Furthermore, melatonin reversed the alterations of protein expression caused by high glucose treatment. Our results concluded that melatonin alleviates high glucose-induced apoptosis in Schwann cells through mitigating mitochondrial-related oxidative stress and the alterations of Bcl-2, NF-κB, mTOR and Wnt signalling pathways.
Collapse
Affiliation(s)
- Yee Lian Tiong
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Soi Moi Chye
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
9
|
Melatonin as a master regulator of cell death and inflammation: molecular mechanisms and clinical implications for newborn care. Cell Death Dis 2019; 10:317. [PMID: 30962427 PMCID: PMC6453953 DOI: 10.1038/s41419-019-1556-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022]
Abstract
Melatonin, more commonly known as the sleep hormone, is mainly secreted by the pineal gland in dark conditions and regulates the circadian rhythm of the organism. Its intrinsic properties, including high cell permeability, the ability to easily cross both the blood–brain and placenta barriers, and its role as an endogenous reservoir of free radical scavengers (with indirect extra activities), confer it beneficial uses as an adjuvant in the biomedical field. Melatonin can exert its effects by acting through specific cellular receptors on the plasma membrane, similar to other hormones, or through receptor-independent mechanisms that involve complex molecular cross talk with other players. There is increasing evidence regarding the extraordinary beneficial effects of melatonin, also via exogenous administration. Here, we summarize molecular pathways in which melatonin is considered a master regulator, with attention to cell death and inflammation mechanisms from basic, translational and clinical points of view in the context of newborn care.
Collapse
|
10
|
Phan TX, Malkani RG. Sleep and circadian rhythm disruption and stress intersect in Alzheimer's disease. Neurobiol Stress 2019; 10:100133. [PMID: 30937343 PMCID: PMC6279965 DOI: 10.1016/j.ynstr.2018.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/12/2018] [Accepted: 10/13/2018] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) was discovered and the pathological hallmarks were revealed more than a century ago. Subsequently, many remarkable discoveries and breakthroughs provided us with mechanistic insights into the pathogenesis of AD. The identification of the molecular underpinning of the disease not only provided the framework of AD pathogenesis but also targets for therapeutic inventions. Despite all the initial successes, no effective treatment for AD has emerged yet as all the late stages of clinical trials have failed. Many factors ranging from genetic to environmental factors have been critically appraised as the potential causes of AD. In particular, the role of stress on AD has been intensively studied while the relationship between sleep and circadian rhythm disruption (SCRD) and AD have recently emerged. SCRD has always been thought to be a corollary of AD pathologies until recently, multiple lines of evidence converge on the notion that SCRD might be a contributing factor in AD pathogenesis. More importantly, how stress and SCRD intersect and make their concerted contributions to AD phenotypes has not been reviewed. The goal of this literature review is to examine at multiple levels - molecular, cellular (e.g. microglia, gut microbiota) and holistic - how the interaction between stress and SCRD bi-directionally and synergistically exacerbate AD pathologies and cognitive impairment. AD, in turn, worsens stress and SCRD and forms the vicious cycle that perpetuates and amplifies AD.
Collapse
Affiliation(s)
- Trongha X. Phan
- Department of Neurology, Division of Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Circadian and Sleep Medicine, Northwestern University, Chicago, IL, USA
| | - Roneil G. Malkani
- Department of Neurology, Division of Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Circadian and Sleep Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
11
|
The Role of Interleukin-18, Oxidative Stress and Metabolic Syndrome in Alzheimer's Disease. J Clin Med 2017; 6:jcm6050055. [PMID: 28531131 PMCID: PMC5447946 DOI: 10.3390/jcm6050055] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/06/2017] [Accepted: 05/18/2017] [Indexed: 12/12/2022] Open
Abstract
The role of interleukins (ILs) and oxidative stress (OS) in precipitating neurodegenerative diseases including sporadic Alzheimer's disease (AD), requires further clarification. In addition to neuropathological hallmarks-extracellular neuritic amyloid-β (Aβ) plaques, neurofibrillary tangles (NFT) containing hyperphosphorylated tau and neuronal loss-chronic inflammation, as well as oxidative and excitotoxic damage, are present in the AD brain. The pathological sequelae and the interaction of these events during the course of AD need further investigation. The brain is particularly sensitive to OS, due to the richness of its peroxidation-sensitive fatty acids, coupled with its high oxygen demand. At the same time, the brain lack robust antioxidant systems. Among the multiple mechanisms and triggers by which OS can accumulate, inflammatory cytokines can sustain oxidative and nitrosative stress, leading eventually to cellular damage. Understanding the consequences of inflammation and OS may clarify the initial events underlying AD, including in interaction with genetic factors. Inflammatory cytokines are potential inducers of aberrant gene expression through transcription factors. Susceptibility disorders for AD, including obesity, type-2 diabetes, cardiovascular diseases and metabolic syndrome have been linked to increases in the proinflammatory cytokine, IL-18, which also regulates multiple AD related proteins. The association of IL-18 with AD and AD-linked medical conditions are reviewed in the article. Such data indicates that an active lifestyle, coupled to a healthy diet can ameliorate inflammation and reduce the risk of sporadic AD.
Collapse
|
12
|
Romero A, Ramos E, Patiño P, Oset-Gasque MJ, López-Muñoz F, Marco-Contelles J, Ayuso MI, Alcázar A. Melatonin and Nitrones As Potential Therapeutic Agents for Stroke. Front Aging Neurosci 2016; 8:281. [PMID: 27932976 PMCID: PMC5120103 DOI: 10.3389/fnagi.2016.00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/10/2016] [Indexed: 01/20/2023] Open
Abstract
Stroke is a disease of aging affecting millions of people worldwide, and recombinant tissue-type plasminogen activator (r-tPA) is the only treatment approved. However, r-tPA has a low therapeutic window and secondary effects which limit its beneficial outcome, urging thus the search for new more efficient therapies. Among them, neuroprotection based on melatonin or nitrones, as free radical traps, have arisen as drug candidates due to their strong antioxidant power. In this Perspective article, an update on the specific results of the melatonin and several new nitrones are presented.
Collapse
Affiliation(s)
- Alejandro Romero
- Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid Madrid, Spain
| | - Eva Ramos
- Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid Madrid, Spain
| | - Paloma Patiño
- Paediatric Unit, La Paz University Hospital Madrid, Spain
| | - Maria J Oset-Gasque
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University of Madrid, Ciudad Universitaria Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, Camilo José Cela UniversityMadrid, Spain; Neuropsychopharmacology Unit, "Hospital 12 de Octubre" Research InstituteMadrid, Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC) Madrid, Spain
| | - María I Ayuso
- Neurovascular Research Group, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Sevilla, Spain
| | - Alberto Alcázar
- Department of Investigation, IRYCIS, Hospital Ramón y Cajal, Madrid, Spain
| |
Collapse
|
13
|
Hendaus MA, Jomha FA, Alhammadi AH. Melatonin in the management of perinatal hypoxic-ischemic encephalopathy: light at the end of the tunnel? Neuropsychiatr Dis Treat 2016; 12:2473-2479. [PMID: 27729791 PMCID: PMC5045913 DOI: 10.2147/ndt.s115533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) affects one to three per 1,000 live full-term births and can lead to severe and permanent neuropsychological sequelae, such as cerebral palsy, epilepsy, mental retardation, and visual motor or visual perceptive dysfunction. Melatonin has begun to be contemplated as a good choice in order to diminish the neurological sequelae from hypoxic-ischemic brain injury. Melatonin emerges as a very interesting medication, because of its capacity to cross all physiological barriers extending to subcellular compartments and its safety and effectiveness. The purpose of this commentary is to detail the evidence on the use of melatonin as a neuroprotection agent. The pharmacologic aspects of the drug as well as its potential neuroprotective characteristics in human and animal studies are described in this study. Melatonin seems to be safe and beneficial in protecting neonatal brains from perinatal HIE. Larger randomized controlled trials in humans are required, to implement a long-awaited feasible treatment in order to avoid the dreaded sequelae of HIE.
Collapse
Affiliation(s)
- Mohamed A Hendaus
- Department of Pediatrics, Section of Academic General Pediatrics, Hamad Medical Corporation
- Department of Clinical Pediatrics, Weill-Cornell Medical College, Doha, Qatar
| | - Fatima A Jomha
- School of Pharmacy, Lebanese International University, Khiara, Lebanon
| | - Ahmed H Alhammadi
- Department of Pediatrics, Section of Academic General Pediatrics, Hamad Medical Corporation
- Department of Clinical Pediatrics, Weill-Cornell Medical College, Doha, Qatar
| |
Collapse
|
14
|
Melatonin modulates endoplasmic reticulum stress and Akt/GSK3-beta signaling pathway in a rat model of renal warm ischemia reperfusion. Anal Cell Pathol (Amst) 2015; 2015:635172. [PMID: 26229743 PMCID: PMC4502281 DOI: 10.1155/2015/635172] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Melatonin (Mel) is widely used to attenuate ischemia/reperfusion (I/R) injury in several organs. Nevertheless, the underlying mechanisms remain unclear. This study was conducted to explore the effect of Mel on endoplasmic reticulum (ER) stress, Akt and MAPK cascades after renal warm I/R. Eighteen Wistar rats were randomized into three groups: Sham, I/R, and Mel + I/R. The ischemia period was 60 min followed by 120 min of reperfusion. Mel (10 mg/kg) was administrated 30 min prior to ischemia. The creatinine clearance, MDA, LDH levels, and histopathological changes were evaluated. In addition, Western blot was performed to study ER stress and its downstream apoptosis as well as phosphorylation of Akt, GSK-3β, VDAC, ERK, and P38. Mel decreased cytolysis and lipid peroxidation and improved renal function and morphology compared to I/R group. Parallely, it significantly reduced the ER stress parameters including GRP 78, p-PERK, XBP 1, ATF 6, CHOP, and JNK. Simultaneously, p-Akt level was significantly enhanced and its target molecules GSK-3β and VDAC were inhibited. Furthermore, the ERK and P38 phosphorylation were evidently augmented after Mel administration in comparison to I/R group. In conclusion, Mel improves the recovery of renal function by decreasing ER stress and stimulating Akt pathway after renal I/R injury.
Collapse
|
15
|
Yu J, Bao C, Dong Y, Liu X. Activation of autophagy in rat brain cells following focal cerebral ischemia reperfusion through enhanced expression of Atg1/pULK and LC3. Mol Med Rep 2015; 12:3339-3344. [PMID: 26018745 PMCID: PMC4526088 DOI: 10.3892/mmr.2015.3850] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 04/30/2015] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the activation of Atg1/pULK, and LC3 in the cerebral cortex following focal cerebral ischemia reperfusion (CIR) injury, thereby examining its effect on autophagy in brain cells. Rat CIR models were established using the technique of middle cerebral artery occlusion. The neurological function score, TTC staining and the water content of brain tissue were used to evaluate the CIR model. Levels of autophagy in the brain cells were examined at different time‑points following CIR damage using electron microscopy. Immunohistochemistry and western blot analysis were also used for the qualitative and quantitative detection of levels of Atg1/pULK and LC3 in the cerebral cortex. Autophagy was observed in the early stage of CIR, and the expression of Atg1/pULK and LC3 were observed 1 h following CIR in the rats and reached peak expression levels after12 h, which following which the they gradually decreased. These results suggested Atg1/pULK and LC3 are key in the regulation of autophagy following CIR in the rat brain.
Collapse
Affiliation(s)
- Jingwei Yu
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Cuifen Bao
- Key Laboratory of Molecular Cell Biology and New Drug Development, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yanru Dong
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xia Liu
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
16
|
Huai Y, Dong Y, Xu J, Meng N, Song C, Li W, Lv P. L-3-n-butylphthalide protects against vascular dementia via activation of the Akt kinase pathway. Neural Regen Res 2014; 8:1733-42. [PMID: 25206470 PMCID: PMC4145956 DOI: 10.3969/j.issn.1673-5374.2013.19.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 05/27/2013] [Indexed: 12/25/2022] Open
Abstract
As a neuroprotective drug for the treatment of ischemic stroke, 3-n-butylphthalide, a celery seed extract, has been approved by the State Food and Drug Administration of China as a clinical therapeutic drug for ischemic stroke patients. L-3-n-butylphthalide possesses significant efficacy in the treatment of acute ischemic stroke. The activated Akt kinase pathway can prevent the death of nerve cells and exhibit neuroprotective effects in the brain after stroke. This study provides the hypothesis that l-3-n-butylphthalide has a certain therapeutic effect on vascular dementia, and its mechanism depends on the activation of the Akt kinase pathway. A vascular dementia mouse model was established by cerebral repetitive ischemia/reperfusion, and intragastrically administered l-3-n-butylphthalide daily for 28 consecutive days after ischemia/reperfusion, or 7 consecutive days before ischemia/reperfusion. The Morris water maze test showed significant impairment of spatial learning and memory at 4 weeks after operation, but intragastric administration of l-3-n-butylphthalide, especially pretreatment with l-3-n-butylphthalide, significantly reversed these changes. Thionine staining and western blot analylsis showed that preventive and therapeutic application of l-3-n-butylphthalide can reduce loss of pyramidal neurons in the hippocampal CA1 region and alleviate nerve damage in mice with vascular dementia. In addition, phosphorylated Akt expression in hippocampal tissue increased significantly after l-3-n- butylphthalide treatment. Experimental findings demonstrate that l-3-n-butylphthalide has preventive and therapeutic effects on vascular dementia, and its mechanism may be mediated by upregulation of phosphorylated Akt in the hippocampus.
Collapse
Affiliation(s)
- Yaping Huai
- Department of Neurology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei Province, China
| | - Jing Xu
- Department of Neurology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Nan Meng
- Department of Neurology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Chunfeng Song
- Electron Microscope Center, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Wenbin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China ; Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei Province, China
| |
Collapse
|
17
|
Sutinen EM, Korolainen MA, Häyrinen J, Alafuzoff I, Petratos S, Salminen A, Soininen H, Pirttilä T, Ojala JO. Interleukin-18 alters protein expressions of neurodegenerative diseases-linked proteins in human SH-SY5Y neuron-like cells. Front Cell Neurosci 2014; 8:214. [PMID: 25147500 PMCID: PMC4124869 DOI: 10.3389/fncel.2014.00214] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/16/2014] [Indexed: 12/27/2022] Open
Abstract
Chronic inflammation and oxidative stress (OS) are present in Alzheimer's disease (AD) brains in addition to neuronal loss, Amyloid-β (Aβ) plaques and hyperphosphorylated tau-protein neurofibrillary tangles (NFTs). Previously we showed that levels of the pro-inflammatory cytokine, interleukin-18 (IL-18), are elevated in post-mortem AD brains. IL-18 can modulate the tau kinases, Cdk5 and GSK3β, as well as Aβ-production. IL-18 levels are also increased in AD risk diseases, including type-2 diabetes and obesity. Here, we explored other IL-18 regulated proteins in neuron-like SH-SY5Y cells. Differentiated SH-SY5Y cells, incubated with IL-18 for 24, 48, or 72 h, were analyzed by two-dimensional gel electrophoresis (2D-DIGE). Specific altered protein spots were chosen and identified with mass spectrometry (MS) and verified by western immunoblotting (WIB). IL-18 had time-dependent effects on the SH-SY5Y proteome, modulating numerous protein levels/modifications. We concentrated on those related to OS (DDAH2, peroxiredoxins 2, 3, and 6, DJ-1, BLVRA), Aβ-degradation (MMP14, TIMP2), Aβ-aggregation (Septin-2), and modifications of axon growth and guidance associated, collapsin response mediator protein 2 (CRMP2). IL-18 significantly increased antioxidative enzymes, indicative of OS, and altered levels of glycolytic α- and γ-enolase and multifunctional 14-3-3γ and -ε, commonly affected in neurodegenerative diseases. MMP14, TIMP2, α-enolase and 14-3-3ε, indirectly involved in Aβ metabolism, as well as Septin-2 showed changes that increase Aβ levels. Increased 14-3-3γ may contribute to GSK3β driven tau hyperphosphorylation and CRMP2 Thr514 and Ser522 phosphorylation with the Thr555-site, a target for Rho kinase, showing time-dependent changes. IL-18 also increased caspase-1 levels and vacuolization of the cells. Although our SH-SY5Y cells were not aged, as neurons in AD, our work suggests that heightened or prolonged IL-18 levels can drive protein changes of known relevance to AD pathogenesis.
Collapse
Affiliation(s)
- Elina M Sutinen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland Kuopio, Finland ; Brain Research Unit, Clinical Research Centre, University of Eastern Finland Kuopio, Finland
| | | | - Jukka Häyrinen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland Kuopio, Finland
| | - Irina Alafuzoff
- Rudbecklaboratoriet, Department of Immunology, Genetics and Pathology, Molecular and Morphological Pathology, Uppsala University Uppsala, Sweden
| | - Steven Petratos
- Regenerative Neuroscience and Development Laboratory, Department of Medicine, Central Clinical School, Monash University Prahran, VIC, Australia
| | - Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland Kuopio, Finland ; Department of Neurology, Kuopio University Hospital Kuopio, Finland
| | - Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland Kuopio, Finland ; Brain Research Unit, Clinical Research Centre, University of Eastern Finland Kuopio, Finland ; Department of Neurology, Kuopio University Hospital Kuopio, Finland
| | - Tuula Pirttilä
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland Kuopio, Finland ; Brain Research Unit, Clinical Research Centre, University of Eastern Finland Kuopio, Finland ; Department of Neurology, Kuopio University Hospital Kuopio, Finland
| | - Johanna O Ojala
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland Kuopio, Finland ; Brain Research Unit, Clinical Research Centre, University of Eastern Finland Kuopio, Finland
| |
Collapse
|
18
|
Shu Y, Zhang H, Kang T, Zhang JJ, Yang Y, Liu H, Zhang L. PI3K/Akt signal pathway involved in the cognitive impairment caused by chronic cerebral hypoperfusion in rats. PLoS One 2013; 8:e81901. [PMID: 24339978 PMCID: PMC3858283 DOI: 10.1371/journal.pone.0081901] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 10/17/2013] [Indexed: 11/19/2022] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a common pathophysiological state that usually occurs in conditions such as vascular dementia and Alzheimer's disease, both of which are characterized by cognitive impairment. In previous studies we found that learning capacity and memory were gradually impaired with CCH, which altered the expression of synaptophysin, microtubule associated protein-2, growth associated protein-43, brain-derived neurotrophic factor, nerve growth factor, N-methyl-D-aspartate receptor subunit 1, cAMP response element-binding protein and tau hyperphosphorylation in the hippocampus. However, the molecular basis of cognitive impairment in CCH remains obscure. Here we explore the hypothesis that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signal pathway is involved in this type of cognitive impairment. In order to determine if the expression of PI3K, Akt and phosphorylated Akt (p-Akt) proteins are altered at different stages of CCH with differing levels of cognitive impairment. we performed permanent, bilateral occlusion of the common carotid arteries (2-VO) to induce CCH. Adult male SD rats were randomly divided into sham-operated group, 2-VO 1 week group, 2-VO 4 weeks group and 2-VO 8 weeks group. Behavior tests were utilized to assess cognitive abilities, while western blots were utilized to evaluate protein expression. Rats in the 2-VO groups spent less time exploring novel objects than those in the sham-operated group, and the discrimination ratio of the 2-VO 8 weeks group and the sham-operated group were higher than chance (0.50). Escape latencies in the Morris water maze task in the 2-VO 1 week group were longer than those in the sham-operated group on day 4 and day 5, while escape latencies in the 2-VO 4 weeks group were longer than those in the sham-operated group from day 3 to day 5. Escape latencies in 2-VO 8 weeks group were longer than those in the sham-operated group from day 2 to day 5. NE (northeast) square swimming times in the 2-VO 1 week group, 2-VO 4 weeks group and 2-VO 8 weeks group were shorter than that in the sham-operated group. Western blotting showed that the PI3K expression in the 2-VO 1 week group was lower than that in sham-operated group, while p-Akt expression in the 2-VO 8 weeks group was higher than that in the sham-operated group. There was a linear relationship between the PI3K expression and the discrimination ratio, as well as a linear relationship between the PI3K and NE square swimming time. Thus, we propose that the PI3K/Akt signal pathway is an important cell pathway that is associated with the cognitive impairment following CCH.
Collapse
Affiliation(s)
- Yi Shu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hong Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
- * E-mail:
| | - Tao Kang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun-jian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Yang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Neuroprotective effect of melatonin: a novel therapy against perinatal hypoxia-ischemia. Int J Mol Sci 2013; 14:9379-95. [PMID: 23629670 PMCID: PMC3676788 DOI: 10.3390/ijms14059379] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 01/01/2023] Open
Abstract
One of the most common causes of mortality and morbidity in children is perinatal hypoxia-ischemia (HI). In spite of the advances in neonatology, its incidence is not diminishing, generating a pediatric population that will require an extended amount of chronic care throughout their lifetime. For this reason, new and more effective neuroprotective strategies are urgently required, in order to minimize as much as possible the neurological consequences of this encephalopathy. In this sense, interest has grown in the neuroprotective possibilities of melatonin, as this hormone may help to maintain cell survival through the modulation of a wide range of physiological functions. Although some of the mechanisms by which melatonin is neuroprotective after neonatal asphyxia remain a subject of investigation, this review tries to summarize some of the most recent advances related with its use as a therapeutic drug against perinatal hypoxic-ischemic brain injury, supporting the high interest in this indoleamine as a future feasible strategy for cerebral asphyctic events.
Collapse
|
20
|
Cho EH, Koh PO. Proteomic identification of proteins differentially expressed by melatonin in hepatic ischemia-reperfusion injury. J Pineal Res 2010; 49:349-55. [PMID: 20666976 DOI: 10.1111/j.1600-079x.2010.00799.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hepatic ischemia-reperfusion (I-R) injury induces hepatic dysfunction or failure. Melatonin is a potent free radical scavenger and a strong antioxidant. Although many studies have demonstrated the protective effect of melatonin in hepatic injury, the molecular mechanisms of this protection are unclear. We identified specific proteins that are differentially expressed by melatonin treatment in hepatic I-R injury. Adult mice were subjected to 1 hr of ischemia and 3 hr of reperfusion. Animals were treated with vehicle or melatonin (10 mg/kg, i.p.) 15 min prior to ischemia and just before reperfusion. Serum aspartate aminotransferase and alanine aminotransferase levels were higher in I-R group than in sham-operated group, and these increases were reduced by melatonin treatment. Proteins that were differentially expressed following melatonin treatment during hepatic I-R injury were detected using two-dimensional gel electrophoresis. Hepatic I-R injury induced down-regulation of glyoxalase I, glutaredoxin-3, spermidine synthase, proteasome subunit beta type-4, and dynamin like protein-1 (DLP-1). However, melatonin prevented the reductions in these proteins induced by I-R injury. Among the identified proteins, we focused on DLP-1, which is essential for the maintenance of mitochondrial and endoplasmic reticulum morphology. Western blot analysis confirmed that melatonin prevents the hepatic I-R injury-induced decrease in DLP-1. These results suggest that melatonin protects hepatic cells against hepatic I-R injury and that its protective effects involve the regulation of specific proteins.
Collapse
Affiliation(s)
- Eun-Hae Cho
- Department of Anatomy, College of Veterinary Medicine and Research Instituite of Life Science, Gyeongsang National University, Jinju, South Korea
| | | |
Collapse
|
21
|
Esposito E, Cuzzocrea S. Antiinflammatory activity of melatonin in central nervous system. Curr Neuropharmacol 2010; 8:228-42. [PMID: 21358973 PMCID: PMC3001216 DOI: 10.2174/157015910792246155] [Citation(s) in RCA: 270] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Revised: 04/25/2010] [Accepted: 05/08/2010] [Indexed: 12/15/2022] Open
Abstract
Melatonin is mainly produced in the mammalian pineal gland during the dark phase. Its secretion from the pineal gland has been classically associated with circadian and circanual rhythm regulation. However, melatonin production is not confined exclusively to the pineal gland, but other tissues including retina, Harderian glands, gut, ovary, testes, bone marrow and lens also produce it. Several studies have shown that melatonin reduces chronic and acute inflammation. The immunomodulatory properties of melatonin are well known; it acts on the immune system by regulating cytokine production of immunocompetent cells. Experimental and clinical data showing that melatonin reduces adhesion molecules and pro-inflammatory cytokines and modifies serum inflammatory parameters. As a consequence, melatonin improves the clinical course of illnesses which have an inflammatory etiology. Moreover, experimental evidence supports its actions as a direct and indirect antioxidant, scavenging free radicals, stimulating antioxidant enzymes, enhancing the activities of other antioxidants or protecting other antioxidant enzymes from oxidative damage. Several encouraging clinical studies suggest that melatonin is a neuroprotective molecule in neurodegenerative disorders where brain oxidative damage has been implicated as a common link. In this review, the authors examine the effect of melatonin on several neurological diseases with inflammatory components, including dementia, Alzheimer disease, Parkinson disease, multiple sclerosis, stroke, and brain ischemia/reperfusion but also in traumatic CNS injuries (traumatic brain and spinal cord injury).
Collapse
Affiliation(s)
- Emanuela Esposito
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Italy
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| |
Collapse
|
22
|
Das A, McDowell M, Pava MJ, Smith JA, Reiter RJ, Woodward JJ, Varma AK, Ray SK, Banik NL. The inhibition of apoptosis by melatonin in VSC4.1 motoneurons exposed to oxidative stress, glutamate excitotoxicity, or TNF-alpha toxicity involves membrane melatonin receptors. J Pineal Res 2010; 48:157-69. [PMID: 20082663 PMCID: PMC2862889 DOI: 10.1111/j.1600-079x.2009.00739.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Loss of motoneurons may underlie some of the deficits in motor function associated with the central nervous system (CNS) injuries and diseases. We tested whether melatonin, a potent antioxidant and free radical scavenger, would prevent motoneuron apoptosis following exposure to toxins and whether this neuroprotection is mediated by melatonin receptors. Exposure of VSC4.1 motoneurons to either 50 microm H(2)O(2), 25 microm glutamate (LGA), or 50 ng/mL tumor necrosis factor-alpha (TNF-alpha) for 24 h caused significant increases in apoptosis, as determined by Wright staining and ApopTag assay. Analyses of mRNA and proteins showed increased expression and activities of stress kinases and cysteine proteases and loss of mitochondrial membrane potential during apoptosis. These insults also caused increases in intracellular free [Ca(2+)] and activities of calpain and caspases. Cells exposed to stress stimuli for 15 min were then treated with 200 nm melatonin. Post-treatment of cells with melatonin attenuated production of reactive oxygen species (ROS) and phosphorylation of p38, MAPK, and JNK1, prevented cell death, and maintained whole-cell membrane potential, indicating functional neuroprotection. Melatonin receptors (MT1 and MT2) were upregulated following treatment with melatonin. To confirm the involvement of MT1 and MT2 in providing neuroprotection, cells were post-treated (20 min) with 10 microm luzindole (melatonin receptor antagonist). Luzindole significantly attenuated melatonin-induced neuroprotection, suggesting that melatonin worked, at least in part, via its receptors to prevent VSC4.1 motoneuron apoptosis. Results suggest that neuroprotection rendered by melatonin to motoneurons is receptor mediated and melatonin may be an effective neuroprotective agent to attenuate motoneuron death in CNS injuries and diseases.
Collapse
Affiliation(s)
- Arabinda Das
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
| | - Misty McDowell
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
| | - Matthew J Pava
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas, San Antonio, TX 78229, USA
| | - John J. Woodward
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
| | - Abhay K. Varma
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Naren L. Banik
- Department of Neurosciences (Division of Neurology), Medical University of South Carolina, Charleston, SC 29425, USA
- Correspondence to: Naren L. Banik, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425. Phone: (843) 792-8570; Fax: (843) 792-8626; Naren L. Banik ()
| |
Collapse
|
23
|
Abstract
Neonatal brain injury is an important cause of death and disability, with pathways of oxidant stress, inflammation, and excitotoxicity that lead to damage that progresses over a long period of time. Therapies have classically targeted individual pathways during early phases of injury, but more recent therapies such as growth factors may also enhance cell proliferation, differentiation, and migration over time. More recent evidence suggests combined therapy may optimize repair, decreasing cell injury while increasing newly born cells.
Collapse
Affiliation(s)
| | - Donna M. Ferriero
- Department of Pediatrics; University of California, San Francisco (FFG, DMF)
- Department of Neurology; University of California, San Francisco (DMF)
| |
Collapse
|
24
|
Abstract
Melatonin plays a neuroprotective role in models of neurodegenerative diseases. However, the molecular mechanisms underlying neuroprotection by melatonin are not well understood. Apoptotic cell death in the central nervous system is a feature of neurodegenerative diseases. The intrinsic and extrinsic apoptotic pathways and the antiapoptotic survival signal pathways play critical roles in neurodegeneration. This review summarizes the reports to date showing inhibition by melatonin of the intrinsic apoptotic pathways in neurodegenerative diseases including stroke, Alzheimer disease, Parkinson disease, Huntington disease, and amyotrophic lateral sclerosis. Furthermore, the activation of survival signal pathways by melatonin in neurodegenerative diseases is discussed.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Wang X, Figueroa BE, Stavrovskaya IG, Zhang Y, Sirianni AC, Zhu S, Day AL, Kristal BS, Friedlander RM. Methazolamide and melatonin inhibit mitochondrial cytochrome C release and are neuroprotective in experimental models of ischemic injury. Stroke 2009; 40:1877-85. [PMID: 19299628 DOI: 10.1161/strokeaha.108.540765] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The identification of a neuroprotective drug for stroke remains elusive. Given that mitochondria play a key role both in maintaining cellular energetic homeostasis and in triggering the activation of cell death pathways, we evaluated the efficacy of newly identified inhibitors of cytochrome c release in hypoxia/ischemia induced cell death. We demonstrate that methazolamide and melatonin are protective in cellular and in vivo models of neuronal hypoxia. METHODS The effects of methazolamide and melatonin were tested in oxygen/glucose deprivation-induced death of primary cerebrocortical neurons. Mitochondrial membrane potential, release of apoptogenic mitochondrial factors, pro-IL-1beta processing, and activation of caspase -1 and -3 were evaluated. Methazolamide and melatonin were also studied in a middle cerebral artery occlusion mouse model. Infarct volume, neurological function, and biochemical events were examined in the absence or presence of the 2 drugs. RESULTS Methazolamide and melatonin inhibit oxygen/glucose deprivation-induced cell death, loss of mitochondrial membrane potential, release of mitochondrial factors, pro-IL-1beta processing, and activation of caspase-1 and -3 in primary cerebrocortical neurons. Furthermore, they decrease infarct size and improve neurological scores after middle cerebral artery occlusion in mice. CONCLUSIONS We demonstrate that methazolamide and melatonin are neuroprotective against cerebral ischemia and provide evidence of the effectiveness of a mitochondrial-based drug screen in identifying neuroprotective drugs. Given the proven human safety of melatonin and methazolamide, and their ability to cross the blood-brain-barrier, these drugs are attractive as potential novel therapies for ischemic injury.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurosurgery, Neuroapoptosis Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chen Z, Chua CC, Gao J, Chua KW, Ho YS, Hamdy RC, Chua BH. Prevention of ischemia/reperfusion-induced cardiac apoptosis and injury by melatonin is independent of glutathione peroxdiase 1. J Pineal Res 2009; 46:235-41. [PMID: 19141089 PMCID: PMC2752734 DOI: 10.1111/j.1600-079x.2008.00654.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Free-radical generation is one of the primary causes of myocardial ischemia/reperfusion (I/R) injury. Melatonin is an efficient free-radical scavenger and induces the expression of antioxidant enzymes. We have previously shown that melatonin can prevent free-radical-induced myocardial injury. To date, the mechanism underlying melatonin's cardioprotective effect is not clear. In this study, we assessed the ability of melatonin to protect against I/R injury in mice deficient in glutathione peroxidase 1 (Gpx1). Mice hearts were subjected to 40 min of global ischemia in vitro followed by 45 min of reperfusion. Myocardial I/R injury (expressed as % of recovery of left ventricular developed pressure x heart rate) was exacerbated in mice deficient in Gpx1 (51 +/- 3% for Gpx1+/+ mice versus 31 +/- 6% for Gpx1(-/-) mice, P < 0.05). Administration of melatonin for 30 min protected against I/R injury in both Gpx1+/+ mice (72 +/- 4.8%) and Gpx1(-/-) mice (63 +/- 4.7%). This protection was accompanied by a significant improvement in left ventricular end-diastolic pressure and a twofold decrease in lactate dehydrogenase (LDH) level released from melatonin-treated hearts. In another set of experiments, mice were subjected to 50 min of ligation of the left descending anterior coronary artery in vivo followed by 4 hr of reperfusion. The infarct sizes, expressed as the percentage of the area at risk, were significantly larger in Gpx1(-/-) mice than in Gpx1+/+ mice (75 +/- 9% versus 54 +/- 6%, P < 0.05) and were reduced significantly in melatonin-treated mice (31 +/- 3.7% Gpx1(-/-) mice and 33 +/- 6.0% Gpx1+/+ mice). In hearts subjected to 30 min of coronary artery occlusion followed by 3 hr of reperfusion, melatonin-treated hearts had significantly fewer in situ oligo ligation-positive myocytes and less protein nitration. Our results demonstrate that the cardioprotective function of melatonin is independent of Gpx1.
Collapse
Affiliation(s)
- Zhongyi Chen
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Chu C. Chua
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Jinping Gao
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Kao-Wei Chua
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences and Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, MI
| | - Ronald C. Hamdy
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Balvin H.L. Chua
- Cecile Cox Quillen Laboratory of Geriatric Research, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Correspondence to Dr. Balvin H.L. Chua, James H. Quillen College of Medicine, East Tennessee State University, Box 70432, Johnson City, TN 37614, Phone: (423) 926-1171 Ext. 7674, Fax: (423) 979-3408,
| |
Collapse
|
27
|
Lin HW, Lee EJ. Effects of melatonin in experimental stroke models in acute, sub-acute, and chronic stages. Neuropsychiatr Dis Treat 2009; 5:157-62. [PMID: 19557110 PMCID: PMC2695239 DOI: 10.2147/ndt.s4815] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Melatonin (n-acetyl-5-methoxy-tryptamine), a naturally occurring indole produced mainly by the pineal gland, is a well known antioxidant. Stroke (cerebral ischemia) is the second leading cause of death worldwide. To date, however, effective and safe treatment for stroke remains unavailable. Melatonin is both lipid- and water-soluble and readily crosses the blood-brain barrier (BBB). Increasing evidence has shown that, in animal stroke models, administering melatonin significantly reduces infarct volume, edema, and oxidative damage and improves electrophysiological and behavioral performance. Here, we reviewed studies that assess effects of melatonin on cerebral ischemia in acute, sub-acute, and chronic stages. In addition to its potent antioxidant properties, melatonin exerts antiapoptotic, antiexcitotoxic, anti-inflammatory effects and promotes mitochondrial functions in animals with cerebral ischemia. Given that melatonin shows almost no toxicity to humans and possesses multifaceted protective capacity against cerebral ischemia, it is valuable to consider using melatonin in clinical trials on patients suffering from stroke.
Collapse
Affiliation(s)
- Hsiao-Wen Lin
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Medical Center and Medical School, Tainan, Taiwan
| | | |
Collapse
|
28
|
KOH PO. Melatonin Attenuates the Cerebral Ischemic Injury via the MEK/ERK/p90RSK/Bad Signaling Cascade. J Vet Med Sci 2008; 70:1219-23. [DOI: 10.1292/jvms.70.1219] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Phil-Ok KOH
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University
| |
Collapse
|